Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
PLoS One ; 11(10): e0162307, 2016.
Article in English | MEDLINE | ID: mdl-27695040

ABSTRACT

Full length TrkC (TrkC-FL) is a receptor tyrosine kinase whose mRNA can be spliced to a truncated TrkC.T1 isoform lacking the kinase domain. Neurotrophin-3 (NT-3) activates TrkC-FL to maintain motor neuron health and function and TrkC.T1 to produce neurotoxic TNF-α; hence resulting in opposing pathways. In mouse and human ALS spinal cord, the reduction of miR-128 that destabilizes TrkC.T1 mRNA results in up-regulated TrkC.T1 and TNF-α in astrocytes. We exploited conformational differences to develop an agonistic mAb 2B7 that selectively activates TrkC-FL, to circumvent TrkC.T1 activation. In mouse ALS, 2B7 activates spinal cord TrkC-FL signals, improves spinal cord motor neuron phenotype and function, and significantly prolongs life-span. Our results elucidate biological paradoxes of receptor isoforms and their role in disease progression, validate the concept of selectively targeting conformational epitopes in naturally occurring isoforms, and may guide the development of pro-neuroprotective (TrkC-FL) and anti-neurotoxic (TrkC.T1) therapeutic strategies.


Subject(s)
Amyotrophic Lateral Sclerosis/physiopathology , Receptor, trkC/physiology , Amyotrophic Lateral Sclerosis/drug therapy , Animals , Antibodies, Monoclonal/immunology , Astrocytes/physiology , Disease Models, Animal , Humans , Mice , MicroRNAs/physiology , Motor Neurons/drug effects , Motor Neurons/physiology , Nerve Growth Factors/physiology , Neuroprotective Agents/therapeutic use , Protein Conformation , Protein Isoforms/physiology , Rats , Receptor, trkC/drug effects , Receptor, trkC/immunology , Tumor Necrosis Factor-alpha/physiology
2.
Cell Microbiol ; 15(8): 1357-66, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23414299

ABSTRACT

Chronic Chagas cardiomyopathy (CCC), caused by the obligate intracellular protozoan parasite Trypanosoma cruzi, is a major cause of morbidity and mortality in Latin America. CCC begins when T. cruzi enters cardiac cells for intracellular multiplication and differentiation, a process that starts with recognition of host-cell entry receptors. However, the nature of these surface molecules and corresponding parasite counter-receptor(s) is poorly understood. Here we show that antibodies against neurotrophin (NT) receptor TrkC, but not against family members TrkA and TrkB, prevent T. cruzi from invading primary cultures of cardiomyocytes and cardiac fibroblasts. Invasion is also selectively blocked by the TrkC ligand NT-3, and by antagonists of Trk autophosphorylation and downstream signalling. Therefore, these results indicate that T. cruzi gets inside cardiomyocytes and cardiac fibroblasts by activating TrkC preferentially over TrkA. Accordingly, short hairpin RNA interference of TrkC (shTrkC), but not TrkA, selectively prevents T. cruzi from entering cardiac cells. Additionally, T. cruzi parasite-derived neurotrophic factor (PDNF)/trans-sialidase, a TrkC-binding protein, but not family member gp85, blocks entry dose-dependently, underscoring the specificity of PDNF as TrkC counter-receptor in cardiac cell invasion. In contrast to invasion, competitive and shRNA inhibition studies demonstrate that T. cruzi-PDNF recognition of TrkA, but not TrkC on primary cardiomyocytes and the cardiomyocyte cell line H9c2 protects the cells against oxidative stress. Thus, this study shows that T. cruzi via PDNF favours neurotrophin receptor TrkC for cardiac cell entry and TrkA for cardiomyocyte protection against oxidative stress, and suggests a new therapeutic opportunity in PDNF and/or fragments thereof for CCC therapy as entry inhibitors and/or cardioprotection agonists.


Subject(s)
Fibroblasts/parasitology , Myocytes, Cardiac/parasitology , Oxidative Stress/physiology , Receptor, trkA/physiology , Receptor, trkC/physiology , Trypanosoma cruzi/pathogenicity , Animals , Cells, Cultured , Chagas Cardiomyopathy , Disease Models, Animal , Fibroblasts/pathology , Fibroblasts/physiology , Glycoproteins/physiology , Host-Parasite Interactions/physiology , Mice , Mice, Inbred C57BL , Myocytes, Cardiac/pathology , Myocytes, Cardiac/physiology , Neuraminidase/physiology , RNA, Small Interfering/pharmacology , Receptor, trkC/antagonists & inhibitors , Receptor, trkC/drug effects , Trypanosoma cruzi/physiology
3.
J Comp Neurol ; 520(11): 2459-74, 2012 Aug 01.
Article in English | MEDLINE | ID: mdl-22237931

ABSTRACT

(±)3,4-Methylenedioxymethamphetamine (MDMA), a widely used drug of abuse, rapidly reduces serotonin levels in the brain when ingested or administered in sufficient quantities, resulting in deficits in complex route-based learning, spatial learning, and reference memory. Neurotrophins are important for survival and preservation of neurons in the adult brain, including serotonergic neurons. In this study, we examined the effects of MDMA on the expression of brain-derived neurotrophic factor (BDNF) and neurotrophin-3 (NT-3) and their respective high-affinity receptors, tropomyosin receptor kinase (trk)B and trkC, in multiple regions of the rat brain. A serotonergic-depleting dose of MDMA (10 mg/kg × 4 at 2-hour intervals on a single day) was administered to adult Sprague-Dawley rats, and brains were examined 1, 7, or 24 hours after the last dose. Messenger RNA levels of BDNF, NT-3, trkB, and trkC were analyzed by using in situ hybridization with cRNA probes. The prefrontal cortex was particularly vulnerable to MDMA-induced alterations in that BDNF, NT-3, trkB, and trkC mRNAs were all upregulated at multiple time points. MDMA-treated animals had increased BDNF expression in the frontal, parietal, piriform, and entorhinal cortices, increased NT-3 expression in the anterior cingulate cortex, and elevated trkC in the entorhinal cortex. In the nigrostriatal system, BDNF expression was upregulated in the substantia nigra pars compacta, and trkB was elevated in the striatum in MDMA-treated animals. Both neurotrophins and trkB were differentially regulated in several regions of the hippocampal formation. These findings suggest a possible role for neurotrophin signaling in the learning and memory deficits seen following MDMA treatment.


Subject(s)
Brain-Derived Neurotrophic Factor/drug effects , Cerebral Cortex/drug effects , Hallucinogens/pharmacology , N-Methyl-3,4-methylenedioxyamphetamine/pharmacology , Neurotrophin 3/drug effects , Animals , Body Temperature/drug effects , Brain-Derived Neurotrophic Factor/genetics , Brain-Derived Neurotrophic Factor/metabolism , Cerebral Cortex/metabolism , Corticosterone/blood , Hippocampus/drug effects , Hippocampus/metabolism , Male , Neostriatum/drug effects , Neostriatum/metabolism , Neurotrophin 3/genetics , Neurotrophin 3/metabolism , RNA, Messenger/analysis , Random Allocation , Rats , Rats, Sprague-Dawley , Receptor, trkB/drug effects , Receptor, trkB/genetics , Receptor, trkB/metabolism , Receptor, trkC/drug effects , Receptor, trkC/genetics , Receptor, trkC/metabolism , Serotonin Agents/pharmacology , Substantia Nigra/drug effects , Substantia Nigra/metabolism , Time Factors , Tyrosine 3-Monooxygenase/drug effects , Tyrosine 3-Monooxygenase/metabolism
4.
Glia ; 49(2): 197-210, 2005 Jan 15.
Article in English | MEDLINE | ID: mdl-15390094

ABSTRACT

The NO-cGMP pathway has emerged as a neuroprotective signaling system involved in communication between neurons and glia. We have previously shown that axotomy or nerve growth factor (NGF)-deprivation of dorsal root ganglion (DRG) neurons leads to increased production of NO and at the same time an increase in cGMP production in their satellite glia cells. Blockade of NO or its receptor, the cGMP synthesizing enzyme soluble guanylate cyclase (sGC), results in apoptosis of neurons and glia. We now show that co-culture of neonatal DRG neurons with either Schwann cells pre-treated with an NO donor or a membrane-permeant cGMP analogue; or neurons maintained in the medium from Schwann cell cultures treated in the same way, prevents neuronal apoptosis. Both NO donor and cGMP treatment of Schwann cells results in synthesis of NGF and NT3. Furthermore, if the Schwann cells are previously infected with adenoviral vectors expressing a dominant negative sGC mutant transgene, treatment of these Schwann cells with an NO donor now fails to prevent neuronal apoptosis. Schwann cells treated in this way also fail to express neither cGMP nor neurotrophins. These findings suggest NO-sGC-cGMP-mediated NGF and NT3 synthesis by Schwann cells protect neurons.


Subject(s)
Cyclic GMP/metabolism , Neuroglia/metabolism , Neurons/metabolism , Neuroprotective Agents/metabolism , Nitric Oxide/metabolism , Peripheral Nervous System/metabolism , Animals , Animals, Newborn , Apoptosis/drug effects , Apoptosis/physiology , Cell Communication/physiology , Cells, Cultured , Coculture Techniques , Culture Media, Conditioned/pharmacology , Cyclic GMP/analogs & derivatives , Cyclic GMP/pharmacology , Cytoprotection , Ganglia, Spinal/drug effects , Ganglia, Spinal/metabolism , Guanylate Cyclase , Nerve Degeneration/metabolism , Nerve Degeneration/prevention & control , Neurons, Afferent/drug effects , Neurons, Afferent/metabolism , Nitric Oxide Donors/pharmacology , Peripheral Nervous System/cytology , Rats , Rats, Wistar , Receptor, Nerve Growth Factor/drug effects , Receptor, Nerve Growth Factor/metabolism , Receptor, trkC/drug effects , Receptor, trkC/metabolism , Receptors, Cytoplasmic and Nuclear/genetics , Schwann Cells/drug effects , Schwann Cells/metabolism , Signal Transduction/genetics , Signal Transduction/physiology , Soluble Guanylyl Cyclase , Transfection
5.
Int J Immunopathol Pharmacol ; 17(2): 157-64, 2004.
Article in English | MEDLINE | ID: mdl-15171816

ABSTRACT

Compounds possessing neurotrophic properties may represent a possible treatment for neurodegenerative disorders such as amyotrophic lateral sclerosis. Xaliproden (SR57746A), an orally-active non-peptide compound, which has been found to exhibit neurotrophic effects in vitro and in vivo, increased the lifespan and delayed the progression of the motor neuron degeneration in PMN mice. We have used a quantitative reverse transcription/polymerase chain reaction amplification technique to study the regulation of neurotrophin mRNA and trk mRNA expression in PMN mice. NGF and NT-3 mRNA are downregulated in PMN mice. These deficiencies can be overcome by a treatment with xaliproden. Such an effect could contribute to neurotrophic effects of xaliproden in vivo and in vitro.


Subject(s)
Naphthalenes/pharmacology , Nerve Growth Factors/biosynthesis , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/metabolism , Pyridines/pharmacology , RNA, Messenger/biosynthesis , Animals , Brain Chemistry/genetics , Brain-Derived Neurotrophic Factor/biosynthesis , DNA Primers , Female , Glyceraldehyde-3-Phosphate Dehydrogenases/metabolism , Male , Mice , Nerve Growth Factor/biosynthesis , Nerve Growth Factor/drug effects , Nerve Growth Factors/genetics , Neurotrophin 3/biosynthesis , Neurotrophin 3/genetics , RNA, Messenger/genetics , Receptor, trkA/biosynthesis , Receptor, trkA/genetics , Receptor, trkC/biosynthesis , Receptor, trkC/drug effects , Receptors, Nerve Growth Factor/biosynthesis , Receptors, Nerve Growth Factor/drug effects , Reverse Transcriptase Polymerase Chain Reaction , Spinal Cord/metabolism
6.
Glia ; 45(2): 111-23, 2004 Jan 15.
Article in English | MEDLINE | ID: mdl-14730705

ABSTRACT

Several studies have demonstrated the potential of olfactory ensheathing cells for the repair of central and peripheral nerve injury. However, the majority of these studies have been performed with olfactory ensheathing cells derived from the olfactory bulbs, situated inside the skull. A more clinically relevant source of olfactory ensheathing cells is the olfactory mucosa, located in the nose. To be successful, an autologous transplant of nasal ensheathing glia would require a large number of purified cells. To address this issue, we have focused our research on three neurotrophic factors, namely nerve growth factor (NGF), brain-derived neurotrophic factor (BDNF), and neurotrophin 3 (NT3). We show here that their respective receptors, TrkA, TrkB, TrkC, as well as p75(NTR) (the low affinity NGF receptor), are expressed in vitro by the nasal ensheathing cells; the three neurotrophins promote purification and proliferation of these glial cells, with an optimal concentration of 50 ng/ml; and human ensheathing cells can be easily biopsied and highly purified using a serum-free medium supplemented with NT3. This technique opens the door for clinical trials in which nasal ensheathing cells will be autotransplanted in humans suffering from nerve injury.


Subject(s)
Cell Culture Techniques/methods , Neuroglia/drug effects , Neurotrophin 3/pharmacology , Olfactory Mucosa/cytology , Adult , Aged , Animals , Biopsy/methods , Brain-Derived Neurotrophic Factor/pharmacology , Brain-Derived Neurotrophic Factor/therapeutic use , Cell Separation , Cells, Cultured , Culture Media, Serum-Free/pharmacology , Female , Glial Fibrillary Acidic Protein/metabolism , Humans , Male , Middle Aged , Nerve Growth Factor/pharmacology , Nerve Growth Factor/therapeutic use , Nerve Regeneration/physiology , Neuroglia/cytology , Neuroglia/transplantation , Neurotrophin 3/therapeutic use , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Receptor, Nerve Growth Factor , Receptor, trkA/drug effects , Receptor, trkA/genetics , Receptor, trkA/metabolism , Receptor, trkB/drug effects , Receptor, trkB/genetics , Receptor, trkB/metabolism , Receptor, trkC/drug effects , Receptor, trkC/genetics , Receptor, trkC/metabolism , Receptors, Nerve Growth Factor/drug effects , Receptors, Nerve Growth Factor/genetics , Receptors, Nerve Growth Factor/metabolism , S100 Proteins/metabolism , Transplantation, Autologous/methods
7.
J Cell Biochem ; 88(5): 865-72, 2003 Apr 01.
Article in English | MEDLINE | ID: mdl-12616526

ABSTRACT

The various members of the Trk tyrosine kinase family and p75 neurotrophin receptor (p75(NTR)) have been identified as signaling receptors for the structurally related members of the neurotrophins (NT) family. We have previously reported that NT treatment of murine and human brain-metastatic melanoma cells affects their invasive capacities and increases the production of extracellular-matrix degradative enzymes. These cells express aberrant levels of functional p75(NTR) and TrkC, the putative high-affinity receptor for the neurotrophin NT-3. Here we demonstrate that, by using sensitive immune-complex kinase assays in human brain-metastatic (70W) melanoma cells, TrkC receptors associate with a kinase activity exhibiting a dose-dependent susceptibility to inhibition by the purine-analogs 6-thioguanine and 2-aminopurine. The activity of this purine-analog-sensitive kinase (PASK) was induced by NT-3 in a time-dependent fashion, phosphorylating exogenous myelin basic protein (MBP) but not denatured enolase. It is similar to the one reported to relate with p75(NTR) and TrkA receptors and stimulated by the prototypic NT, nerve growth factor. Thus, PASKs may represent unique signaling components common to NT receptors that could engage joint downstream signaling effectors in brain-metastatic melanoma.


Subject(s)
Brain Neoplasms/metabolism , Melanoma/metabolism , Protein Kinases/metabolism , Protein Serine-Threonine Kinases/metabolism , 2-Aminopurine/pharmacology , Cell Line, Tumor , Dose-Response Relationship, Drug , Humans , Neoplasm Metastasis , Neurotrophin 3/pharmacology , Protein Kinases/analysis , Protein Kinases/chemistry , Protein Serine-Threonine Kinases/analysis , Protein Serine-Threonine Kinases/chemistry , Receptor, Nerve Growth Factor , Receptor, trkC/chemistry , Receptor, trkC/drug effects , Receptor, trkC/metabolism , Receptors, Nerve Growth Factor/drug effects , Receptors, Nerve Growth Factor/metabolism , Signal Transduction , Thioguanine/pharmacology , Time Factors
8.
Neurosci Lett ; 336(1): 9-12, 2003 Jan 09.
Article in English | MEDLINE | ID: mdl-12493590

ABSTRACT

Neurotrophic factors maintain and modulate neuron function in adults. We tested the hypothesis that neurotrophic factors rapidly alter intracellular calcium concentrations, thereby affecting neuron excitability. The majority of rat nodose neurons express TrkA, TrkB and TrkC receptor after 1 day in culture. Addition of nerve growth factor, brain derived neurotrophic factor or glial derived neurotrophic factor increased cytosolic calcium in about one third of the neurons within less than 10 min. This increase was due to calcium release from intracellular stores and could be blocked by the tyrosine kinase inhibitor K252a. The rapid effect of neurotrophic factors suggests a role of these molecules in the early response after inflammation as potential mediators for sensitization of afferent neurons.


Subject(s)
Calcium/metabolism , Homeostasis/drug effects , Nerve Growth Factors/pharmacology , Neurons/drug effects , Visceral Afferents/drug effects , Animals , Cells, Cultured , Fura-2 , Glial Cell Line-Derived Neurotrophic Factor , Homeostasis/physiology , Male , Nerve Growth Factor/pharmacology , Neurons/metabolism , Nodose Ganglion/drug effects , Nodose Ganglion/metabolism , Rats , Rats, Sprague-Dawley , Receptor, trkA/drug effects , Receptor, trkA/metabolism , Receptor, trkB/drug effects , Receptor, trkB/metabolism , Receptor, trkC/drug effects , Receptor, trkC/metabolism , Time Factors , Visceral Afferents/metabolism
9.
Neuroscience ; 115(4): 1295-308, 2002.
Article in English | MEDLINE | ID: mdl-12453498

ABSTRACT

Neurotrophin-3 (NT-3), a member of the neurotrophin family of neurotrophic factors, is important for cell survival, axonal growth and neuronal plasticity. Epileptiform activation can regulate the expression of neurotrophins, and increases or decreases in neurotrophins can affect both epileptogenesis and seizure-related axonal growth. Interestingly, the expression of nerve growth factor and brain-derived neurotrophic factor is rapidly up-regulated following seizures, while NT-3 mRNA remains unchanged or undergoes a delayed down-regulation, suggesting that NT-3 might have a different function in epileptogenesis. In the present study, we demonstrate that continuous intraventricular infusion of NT-3 in the absence of kindling triggers mossy fiber sprouting in the inner molecular layer of the dentate gyrus and the stratum oriens of the CA3 region. Furthermore, despite this NT-3-related sprouting effect, continuous infusion of NT-3 retards the development of behavioral seizures and inhibits kindling-induced mossy fiber sprouting in the inner molecular layer of the dentate gyrus. We also show that prolonged infusion of NT-3 leads to a decrease in kindling-induced Trk phosphorylation and a down-regulation of the high-affinity Trk receptors, TrkA and TrkC, suggesting an involvement of both cholinergic nerve growth factor receptors and hippocampal NT-3 receptors in these effects. Our results demonstrate an important inhibitory role for NT-3 in seizure development and seizure-related synaptic reorganization.


Subject(s)
Epilepsy/metabolism , Growth Cones/metabolism , Kindling, Neurologic/metabolism , Mossy Fibers, Hippocampal/metabolism , Neuronal Plasticity/physiology , Neurotrophin 3/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Animals , Cell Count , Cytochrome c Group/pharmacology , Drug Administration Schedule , Epilepsy/drug therapy , Epilepsy/physiopathology , Growth Cones/drug effects , Kindling, Neurologic/drug effects , Male , Molecular Weight , Mossy Fibers, Hippocampal/drug effects , Mossy Fibers, Hippocampal/growth & development , Neuronal Plasticity/drug effects , Neuropil/cytology , Neuropil/drug effects , Neurotrophin 3/pharmacology , Phosphorylation/drug effects , Rats , Rats, Long-Evans , Receptor Protein-Tyrosine Kinases/drug effects , Receptor, trkA/drug effects , Receptor, trkA/metabolism , Receptor, trkB/drug effects , Receptor, trkB/metabolism , Receptor, trkC/drug effects , Receptor, trkC/metabolism , Seizures/drug therapy , Seizures/metabolism , Seizures/physiopathology
10.
J Neurosci Res ; 70(3): 335-9, 2002 Nov 01.
Article in English | MEDLINE | ID: mdl-12391593

ABSTRACT

Effects of 4-methycatechol (4MC), a potent stimulator of nerve growth factor and brain-derived neurotrophic factor (BDNF) synthesis, on phosphorylation of cellular molecules in cultured rat cortical neurons were examined. 4MC stimulated tyrosine phosphorylation of various proteins of molecular weight from 10-300 kDa including Trks, which are high-affinity neurotrophin receptors. Moreover, 4MC enhanced the phosphorylation of serine 133 of mitogen-activated protein kinase (MAPK/ERK) in a dose-dependent manner. Pretreatment of cultures with PD98059, a selective inhibitor of MAPK kinase (MEK-1), inhibited 4MC-induced phosphorylation of ERKs, demonstrating MEK-1-mediated activation. Therefore, it seems that 4MC triggered the phosphorylation of Trks, resulting in the activation of the subsequent MAPK/ERK signal cascade, or perhaps the involvement of BDNF action as 4MC can stimulate neuronal BDNF synthesis. The phosphorylation of MAPK/ERK was unaffected, however, in the presence of cycloheximide, a protein synthesis inhibitor, and K252a, a selective inhibitor of Trks, suggesting that the effect of newly synthesized BDNF was negligible on this event, and that primary sites of 4MC actions are not limited only to Trks. These results suggest that 4MC primarily activates multiple signal transduction molecules such as tyrosine kinases, including Trks. A significant increase in the survival rate of cortical neurons in the presence of 10 or 100 nM 4MC supported this idea, because the concentrations were much lower than those for stimulation of BDNF synthesis. Our results strongly suggest that the neurotrophic actions of 4MC found so far are mediated predominantly by direct activation of some intracellular signals including MAPK/ERK rather than by neurotrophin synthesis.


Subject(s)
Aging/metabolism , Catechols/pharmacology , Cerebral Cortex/metabolism , MAP Kinase Signaling System/physiology , Neurons/metabolism , Neuroprotective Agents/pharmacology , Receptors, Nerve Growth Factor/metabolism , Aging/drug effects , Animals , Brain-Derived Neurotrophic Factor/metabolism , Brain-Derived Neurotrophic Factor/pharmacology , Brain-Derived Neurotrophic Factor/therapeutic use , Cell Survival/drug effects , Cell Survival/physiology , Cells, Cultured , Cerebral Cortex/drug effects , Cerebral Cortex/physiopathology , Enzyme Inhibitors/pharmacology , Female , Fetus , MAP Kinase Signaling System/drug effects , Neurons/drug effects , Phosphorylation/drug effects , Pregnancy , Rats , Receptor, trkA/drug effects , Receptor, trkA/metabolism , Receptor, trkB/drug effects , Receptor, trkB/metabolism , Receptor, trkC/drug effects , Receptor, trkC/metabolism , Receptors, Nerve Growth Factor/drug effects
11.
Mol Cell Neurosci ; 20(1): 2-12, 2002 May.
Article in English | MEDLINE | ID: mdl-12056835

ABSTRACT

Neurotrophins and cell adhesion molecules regulate axon guidance, but their potential coordinate interactions are not well defined. In particular, it has been difficult to define the role of signaling from different surface molecules in neurotrophin-induced axon growth because of the strong dependence of embryonic neurons on this class of molecules for survival. We have addressed this issue using Bax deficient neurons, which do not require neurotrophins for survival. The L1 neural cell adhesion molecule and laminin each supported NGF-independent axon growth of cultured sensory neurons from dorsal root ganglia of embryonic Bax(-/-) mice. However, nerve growth factor (NGF) stimulated additional axon growth of sensory neurons on laminin but not on L1 substrates. Inhibition of the small GTPase RhoA by the dominant-negative mutant RhoA(T19N) restored NGF responsiveness of axon growth on L1 to Bax(-/-) neurons. Constitutively activated RhoA(Q63L) did not affect axon growth on L1 but inhibited NGF-stimulated axon growth on laminin. Consistent with the concept that RhoA was downregulated by NGF in neurons on laminin but not L1, the RhoA inhibitor C2IN-C3 toxin stimulated axon growth on L1 in wild-type DRG neurons in NGF. These results demonstrate a novel substrate-dependent regulation of NGF-induced growth of embryonic sensory axons mediated by RhoA GTPase.


Subject(s)
Botulinum Toxins , Cell Differentiation/physiology , Ganglia, Spinal/embryology , Growth Cones/metabolism , Laminin/metabolism , Membrane Glycoproteins/metabolism , Nerve Growth Factor/metabolism , Neural Cell Adhesion Molecules/metabolism , Neurons, Afferent/metabolism , Proto-Oncogene Proteins c-bcl-2 , ADP Ribose Transferases/pharmacology , Animals , Cell Differentiation/drug effects , Cells, Cultured , Drug Interactions/physiology , Female , Fetus , Ganglia, Spinal/cytology , Ganglia, Spinal/metabolism , Growth Cones/drug effects , Growth Cones/ultrastructure , Immunohistochemistry , Laminin/pharmacology , Leukocyte L1 Antigen Complex , Male , Membrane Glycoproteins/pharmacology , Mice , Mice, Knockout , Nerve Growth Factor/pharmacology , Neural Cell Adhesion Molecules/pharmacology , Neurons, Afferent/cytology , Neurons, Afferent/drug effects , Proto-Oncogene Proteins/deficiency , Proto-Oncogene Proteins/genetics , Receptor, trkC/drug effects , Receptor, trkC/metabolism , bcl-2-Associated X Protein , rhoA GTP-Binding Protein/agonists , rhoA GTP-Binding Protein/antagonists & inhibitors , rhoA GTP-Binding Protein/metabolism
12.
Dev Biol ; 226(2): 180-91, 2000 Oct 15.
Article in English | MEDLINE | ID: mdl-11023679

ABSTRACT

Neurotrophin-3 (NT-3) is a member of the neurotrophin family of growth factors, best characterized by its survival- and differentiation-inducing effects on developing neurons bearing the trk C receptor tyrosine kinase. Through analysis of NT-3 and trk C gene-targeted mice we have identified NT-3 as critically regulating cardiac septation, valvulogenesis, and conotruncal formation. Although these defects could reflect cardiac neural crest dysfunction, the expression of NT-3 and trk C by cardiac myocytes prior to neural crest migration prompted analysis of cell-autonomous actions of NT-3 on cardiac myocytes. Retroviral-mediated overexpression of truncated trk C receptor lacking kinase activity was used to inhibit activation of trk C by endogenous NT-3, during early heart development in ovo. During the first week of chicken development, expression of truncated trk C reduced myocyte clone size by more than 60% of control clones. Direct mitogenic actions of NT-3 on embryonic cardiac myocytes were demonstrated by analysis of BrdU incorporation or PCNA immunoreactivity in control and truncated trk C-expressing clones. Inhibition of trk C signaling reduced cardiac myocyte proliferation during the first week of development, but had no effect at later times. These studies demonstrate that endogenous NT-3:trk C signaling regulates cardiac myocyte proliferation during cardiac looping and the establishment of ventricular trabeculation but that myocyte proliferation becomes NT-3 independent during the second week of embryogenesis.


Subject(s)
Fetal Heart/cytology , Myocardium/cytology , Neurotrophin 3/physiology , Receptor, trkC/physiology , Amino Acid Sequence , Animals , Autocrine Communication , Cell Division/drug effects , Chick Embryo , DNA Replication/drug effects , DNA, Complementary/genetics , Defective Viruses/genetics , Fetal Heart/drug effects , Fibroblast Growth Factor 1 , Fibroblast Growth Factor 2/physiology , Genetic Vectors/genetics , Molecular Sequence Data , Myocardium/metabolism , Peptide Fragments/genetics , Peptide Fragments/physiology , Proliferating Cell Nuclear Antigen/analysis , Receptor, trkC/chemistry , Receptor, trkC/deficiency , Receptor, trkC/drug effects , Receptor, trkC/genetics , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/physiology , Retroviridae/genetics
13.
Dev Biol ; 226(2): 220-30, 2000 Oct 15.
Article in English | MEDLINE | ID: mdl-11023682

ABSTRACT

Rohon Beard (RB) cells are embryonic primary sensory neurons that are removed by programmed cell death during larval development in zebrafish. RB somatosensory functions are taken over by neurons of the dorsal root ganglia (DRG), suggesting that RB cell death may be triggered by the differentiation of these ganglia, as has been proposed to be the case in Xenopus. However, here we show that the timing of RB cell death correlates with reduced expression of trkC1, the receptor for neurotrophin NT-3, but not with the appearance of DRG, which differentiate only after most RB cells die. trkC1 is expressed in subpopulations of RB neurons during development, and cell death is initiated only in trkC1-negative neurons, suggesting a role for TrkC1 and its ligand, NT-3, in RB cell survival. In support of this, antibodies that deplete NT-3 induce RB cell death while exogenous application of NT-3 reduces death. In addition, we show that RB cell death can be prevented using a caspase inhibitor, zVADfmk, showing that during normal development, RB cells die by a caspase-dependent programmed cell death pathway possibly triggered by reduced signaling via TrkC1.


Subject(s)
Apoptosis/physiology , Nerve Tissue Proteins/physiology , Neurons, Afferent/cytology , Neurotrophin 3/physiology , Receptor, trkC/physiology , Zebrafish/growth & development , Animals , Apoptosis/drug effects , Caspases/physiology , Cell Differentiation , Cells, Cultured , Ganglia, Spinal/cytology , In Situ Nick-End Labeling , Nerve Tissue Proteins/drug effects , Neurons, Afferent/classification , Neurons, Afferent/drug effects , Neurotrophin 3/pharmacology , Receptor, trkC/drug effects , Signal Transduction , Zebrafish/anatomy & histology
14.
Neuroreport ; 11(11): 2541-5, 2000 Aug 03.
Article in English | MEDLINE | ID: mdl-10943719

ABSTRACT

Although roles of neurotrophin-3 (NT3) and glial cell line-derived neurotrophic factor (GDNF) are suggested for sympathetic neuron development, survival activity of either NT3 or GDNF alone on cultured embryonic rat sympathetic neurons is low (10-20%). We demonstrate that a combination of both NT3 and GDNF exerts a remarkable survival activity (85%). NT3 and GDNF did not affect expression levels of their receptors. However, stimulation with both NT3 and GDNF caused tyrosine-phosphorylation of Ret/GDNF-receptor at a level higher than that caused by GDNF alone. Furthermore, stimulation with both GDNF and NT3 induced an enhanced Thr308-phosphorylation of Akt kinase. These results suggest that the actions of NT3 and GDNF converge at the Ret/GDNF-receptor to enhance survival of the developing sympathetic neurons through activation of the phosphatidylinositol (PI) 3-kinase/Akt pathway.


Subject(s)
Cell Survival/drug effects , Drosophila Proteins , Drug Interactions/physiology , Nerve Growth Factors , Nerve Tissue Proteins/pharmacology , Neurons/drug effects , Neuroprotective Agents/pharmacology , Neurotrophin 3/pharmacology , Proto-Oncogene Proteins/drug effects , Receptor Protein-Tyrosine Kinases/drug effects , Sympathetic Nervous System/drug effects , Animals , Cell Survival/physiology , Cells, Cultured , Fetus , Glial Cell Line-Derived Neurotrophic Factor , Glial Cell Line-Derived Neurotrophic Factor Receptors , Nerve Tissue Proteins/metabolism , Neurons/cytology , Neurons/metabolism , Neuroprotective Agents/metabolism , Neurotrophin 3/metabolism , Phosphatidylinositol 3-Kinases/drug effects , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation/drug effects , Protein Serine-Threonine Kinases/drug effects , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Proto-Oncogene Proteins c-ret , Rats , Rats, Wistar , Receptor Protein-Tyrosine Kinases/metabolism , Receptor, trkC/drug effects , Receptor, trkC/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology , Sympathetic Nervous System/cytology , Sympathetic Nervous System/embryology
SELECTION OF CITATIONS
SEARCH DETAIL
...