Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Mol Cell Biol ; 36(20): 2596-611, 2016 10 15.
Article in English | MEDLINE | ID: mdl-27503856

ABSTRACT

Macropinocytosis is a normal cellular process by which cells internalize extracellular fluids and nutrients from their environment and is one strategy that Ras-transformed pancreatic cancer cells use to increase uptake of amino acids to meet the needs of rapid growth. Paradoxically, in non-Ras transformed medulloblastoma brain tumors, we have shown that expression and activation of the receptor tyrosine kinase TrkA overactivates macropinocytosis, resulting in the catastrophic disintegration of the cell membrane and in tumor cell death. The molecular basis of this uncontrolled form of macropinocytosis has not been previously understood. Here, we demonstrate that the overactivation of macropinocytosis is caused by the simultaneous activation of two TrkA-mediated pathways: (i) inhibition of RhoB via phosphorylation at Ser(185) by casein kinase 1, which relieves actin stress fibers, and (ii) FRS2-scaffolded Src and H-Ras activation of RhoA, which stimulate actin reorganization and the formation of lamellipodia. Since catastrophic macropinocytosis results in brain tumor cell death, improved understanding of the mechanisms involved will facilitate future efforts to reprogram tumors, even those resistant to apoptosis, to die.


Subject(s)
Casein Kinase I/metabolism , Cerebellar Neoplasms/metabolism , Medulloblastoma/metabolism , Pinocytosis , Receptor, trkA/metabolism , rhoB GTP-Binding Protein/metabolism , Actins/metabolism , Cell Death , Cell Line, Tumor , Humans , Phosphorylation , Proto-Oncogene Proteins p21(ras)/metabolism , Serine/metabolism , Signal Transduction , rhoA GTP-Binding Protein/metabolism
2.
J Mol Neurosci ; 49(1): 38-51, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22744634

ABSTRACT

We previously demonstrated that the guanine nucleotide exchange factor, RasGrf1, binds nerve growth factor (NGF)-activated TrkA and facilitates neurotrophin-induced neurite outgrowth in PC12 cells. RasGrf1 can activate both Ras and Rac, via intrinsic Cdc25 and DH domains, respectively, suggesting that the activation of both could contribute to this process. Previous studies have assayed constitutive neurite outgrowth following RasGrf1 over-expression in PC12 cells, in either the absence or presence of ectopic H-Ras, and have suggested an essential role for either Ras or Rac depending on the presence of H-Ras over-expression. In contrast, in this study, we have addressed the mechanism of how RasGrf1 facilitates neurite outgrowth in response to the neurotrophins, NGF and BDNF. Using Ras/Rac activation assays and site-directed RasGrf1 mutants, we find that both Ras and Rac are essential to neurotrophin-induced neurite outgrowth. Moreover, we find that H-Ras over-expression rescues the loss of neurite outgrowth observed with a Rac minus mutant and that RasGrf1 differentially stimulates NGF-dependent activation of Rac or Ras, depending on cell type. Collectively, these studies clarify the mechanism of how RasGrf1 expression facilitates neurotrophin-induced neurite outgrowth. Moreover, they suggest that H-Ras over-expression should be used with caution to measure phenotypic responses.


Subject(s)
Neurites/physiology , Proto-Oncogene Proteins p21(ras)/metabolism , Receptors, Nerve Growth Factor/metabolism , rac GTP-Binding Proteins/metabolism , ras-GRF1/metabolism , Animals , Brain-Derived Neurotrophic Factor/metabolism , HEK293 Cells , Humans , Mice , Mutagenesis, Site-Directed , Mutation , Nerve Growth Factor/metabolism , Neurites/metabolism , PC12 Cells , Proto-Oncogene Proteins p21(ras)/genetics , Rats , Receptor, trkA/metabolism , Receptor, trkB/metabolism , Up-Regulation , ras-GRF1/genetics
3.
J Neurochem ; 121(6): 861-80, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22404429

ABSTRACT

Vesicular transport in neurons plays a vital role in neuronal function and survival. Nesca is a novel protein that we previously identified and herein describe its pattern of expression, subcellular localization and protein-protein interactions both in vitro and in vivo. Specifically, a large proportion of Nesca is in tight association with both actin and microtubule cytoskeletal proteins. Nesca binds to F-actin, microtubules, ßIII and acetylated α-tubulin, but not neurofilaments or the actin-binding protein drebrin, in in vitro-binding assays. Nesca co-immunoprecipitates with kinesin heavy chain (KIF5B) and kinesin light-chain motors as well as with the synaptic membrane precursor protein, syntaxin-1, and is a constituent of the post-synaptic density. Moreover, in vitro-binding assays indicate that Nesca directly binds KIF5B, kinesin light-chain and syntaxin-1. In contrast, Nesca does not co-immunoprecipitate with the kinesin motors KIF1B, KIF3A nor does it bind syntaxin-4 or the synaptosome-associated protein 25 kDa (SNAP-25) in vitro. Nesca expression in neurons is highly punctuate, co-stains with syntaxin-1, and is found in fractions containing markers of early endosomes and Golgi suggesting that it is involved in vesicular transport. Collectively, these data suggest that Nesca functions as an adapter involved in neuronal vesicular transport including vesicles containing soluble N-ethylmaleimide sensitive factor attachment protein receptors that are essential to exocytosis.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Kinesins/metabolism , Neurons/metabolism , Syntaxin 1/metabolism , Adaptor Proteins, Vesicular Transport/metabolism , Animals , Blotting, Western , Cytoskeletal Proteins/metabolism , Cytoskeleton/metabolism , Hippocampus/metabolism , Humans , Immunohistochemistry , Immunoprecipitation , Mice , Neurogenesis/physiology , Post-Synaptic Density/metabolism , Protein Transport/physiology , Synaptic Membranes/metabolism , Transfection
4.
J Neurochem ; 112(4): 882-99, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19943845

ABSTRACT

Ectopic expression of the TrkA receptor tyrosine kinase in tumors of the nervous system can mediate nerve growth factor (NGF)-dependent cell death by apoptosis and /or autophagy. Herein, we demonstrate that TrkA can also induce cell death in medulloblastoma Daoy cells by a caspase-independent mechanism that involves the hyperstimulation of macropinocytosis. Specifically, NGF-stimulates the uptake of AlexaFluor546-dextran into lysosome-associated membrane protein-1 positive vacuoles which fuse with microtubule associated protein light chain 3 (LC3) positive autophagosomes, to form large intracellular vacuoles (> 1 mum), which then fuse with lysotracker positive lysosomes. While LC3 cleavage and the appearance of LC3 positive vacuoles suggest the induction of autophagy, siRNA reduced expression of four proteins essential to autophagy (beclin-1, Atg5, LC3 and Atg9) neither blocks NGF-induced vacuole formation nor cell death. TrkA activated cell death does not require p38, JNK or Erk1/2 kinases but does require activation of class III PI-3 kinase and is blocked by the casein kinase 1 (CK1) inhibitor, D4476. This inhibitor does not interfere with TrkA activation but does block NGF-dependent AlexaFluor546-dextran uptake and CK1 dependent phosphorylation of beta-catenin. Collectively, these data demonstrate that TrkA stimulates cell death by a novel mechanism involving CK1-dependent hyperstimulation of macropinocytosis.


Subject(s)
Autophagy/drug effects , Nerve Growth Factor/pharmacology , Pinocytosis/drug effects , Pinocytosis/physiology , Receptor, trkA/metabolism , Analysis of Variance , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Autophagy-Related Protein 5 , Beclin-1 , Cell Line, Tumor , Cytochromes c/metabolism , Enzyme Inhibitors/pharmacology , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Immunoprecipitation/methods , Lysosomal Membrane Proteins/genetics , Lysosomal Membrane Proteins/metabolism , Lysosomes/drug effects , Lysosomes/ultrastructure , Medulloblastoma/pathology , Medulloblastoma/ultrastructure , Membrane Proteins/genetics , Membrane Proteins/metabolism , Microscopy, Electron, Transmission/methods , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , RNA, Small Interfering/pharmacology , Signal Transduction/drug effects , Time Factors , Transfection/methods
5.
J Neurochem ; 112(4): 924-39, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19943849

ABSTRACT

Fibroblast growth factor (FGF) mediated signaling is essential to many aspects of neural development. Activated FGF receptors signal primarily through the FGF receptor substrate (Frs) adapters, which include Frs2/Frs2alpha and Frs3/Frs2beta. While some studies suggest that Frs3 can compensate for the loss of Frs2 in transfected cells, the lack of an effective Frs3 specific antibody has prevented efforts to determine the role(s) of the endogenous protein. To this end, we have generated a Frs3 specific antibody and have characterized the pattern of Frs3 expression in the developing nervous system, its subcellular localization as well as its biochemical properties. We demonstrate that Frs3 is expressed at low levels in the ventricular zone of developing cortex, between E12 and E15, and it co-localizes with nestin and acetylated alpha-tubulin in radial processes in the ventricular/subventricular zones as well as with betaIII tubulin in differentiated cortical neurons. Subcellular fractionation studies demonstrate that endogenous Frs3 is both soluble and plasma membrane associated while Frs3 expressed in 293T cells associates exclusively with lipid rafts. Lastly, we demonstrate that neuronal Frs3 binds microtubules comparable to the microtubule-associated protein, MAP2, while Frs2 does not. Collectively, these data suggest that neuronal Frs3 functions as a novel microtubule binding protein and they provide the first biochemical evidence that neuronal Frs3 is functionally distinct from Frs2/Frs2alpha.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cell Differentiation/physiology , Cell Movement/physiology , Gene Expression Regulation, Developmental/physiology , Microtubule-Associated Proteins/metabolism , Neurons/physiology , Adaptor Proteins, Signal Transducing/genetics , Animals , Animals, Newborn , Astrocytes/drug effects , Astrocytes/physiology , Cell Differentiation/drug effects , Cell Movement/drug effects , Cells, Cultured , Cerebral Cortex/cytology , Cerebral Cortex/growth & development , Cyclic AMP/pharmacology , Embryo, Mammalian , Gene Expression Regulation, Developmental/drug effects , Glial Fibrillary Acidic Protein/metabolism , Hippocampus , Humans , In Vitro Techniques , Ki-67 Antigen/metabolism , Lysosomal-Associated Membrane Protein 1/metabolism , Membrane Microdomains/metabolism , Mice , Microtubule-Associated Proteins/genetics , Neurons/drug effects , Protein Binding , T-Box Domain Proteins/metabolism , Tubulin/metabolism
6.
Biochim Biophys Acta ; 1763(4): 366-80, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16697063

ABSTRACT

We have investigated the signaling properties of the fibroblast growth factor (FGF) receptor substrate 3 (FRS3), also known as SNT-2 or FRS2beta, in neurotrophin-dependent differentiation in comparison with the related adapter FRS2 (SNT1 or FRS2alpha). We demonstrate that FRS3 binds all neurotrophin Trk receptor tyrosine kinases and becomes tyrosine phosphorylated in response to NGF, BDNF, NT-3 and FGF stimulation in transfected cells and/or primary cortical neurons. Second, the signaling molecules Grb2 and Shp2 bind FRS3 at consensus sites that are highly conserved among FRS family members and that Shp2, in turn, becomes tyrosine phosphorylated. While FRS3 over-expression in PC12 cells neither increases NGF-induced neuritogenesis nor activation of Map kinase/AKT, comparable to previous reports on FRS2, over-expression of a chimeric adapter containing the PH/PTB domains of the insulin receptor substrate (IRS) 2, in place of the PTB domain of FRS3 (IRS2-FRS3) supports insulin-dependent Map kinase activation and neurite outgrowth in PC12 cells. Collectively, these data demonstrate that FRS3 supports ligand-induced Map kinase activation and that the chimeric IRS2-FRS3 adapter is stimulating sufficient levels of activated MapK to support neurite outgrowth in PC12 cells.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Fibroblast Growth Factors/physiology , Nerve Growth Factors/physiology , Receptor Protein-Tyrosine Kinases/metabolism , Amino Acid Sequence , Animals , Humans , Molecular Sequence Data , PC12 Cells , Rats , Receptor, trkA/metabolism , Receptor, trkB/metabolism , Receptor, trkC/metabolism , Receptors, Fibroblast Growth Factor/metabolism , Substrate Specificity/physiology
7.
J Biol Chem ; 280(20): 19461-71, 2005 May 20.
Article in English | MEDLINE | ID: mdl-15753086

ABSTRACT

The human tumorous imaginal disc 1 (TID1) proteins including TID1(L) and TID1(S), members of the DnaJ domain protein family, are involved in multiple intracellular signaling pathways such as apoptosis induction, cell proliferation, and survival. Here we report that TID1 associates with the Trk receptor tyrosine kinases and regulates nerve growth factor (NGF)-induced neurite outgrowth in PC12-derived nnr5 cells. Binding assays and transfection studies showed that the carboxyl-terminal end of TID1 (residues 224-429) bound to Trk at the activation loop (Tyr(P)(683)-Tyr(684)(P)(684) in rat TrkA) and that TID1 was tyrosine phosphorylated by Trk both in yeast and in transfected cells. Moreover endogenous TID1 was also tyrosine phosphorylated by and co-immunoprecipitated with Trk in neurotrophin-stimulated primary rat hippocampal neurons. Overexpression studies showed that both TID1(L) and TID1(S) significantly facilitated NGF-induced neurite outgrowth in TrkA-expressing nnr5 cells possibly through a mechanism involving increased activation of mitogen-activated protein kinase. Consistently knockdown of endogenous TID1, mediated with specific short hairpin RNA, significantly reduced NGF-induced neurite growth in nnr5-TrkA cells. These data provide the first evidence that TID1 is a novel intracellular adaptor that interacts with the Trk receptor tyrosine kinases in an activity-dependent manner to facilitate Trk-dependent intracellular signaling.


Subject(s)
Heat-Shock Proteins/metabolism , Receptor, trkA/metabolism , Animals , Base Sequence , Binding Sites , DNA/genetics , HSP40 Heat-Shock Proteins , Heat-Shock Proteins/chemistry , Heat-Shock Proteins/genetics , Humans , Kinetics , Neurites/metabolism , Neurites/ultrastructure , PC12 Cells , Phosphorylation , RNA Interference , Rats , Receptor, trkA/chemistry , Receptor, trkA/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Signal Transduction , Transfection , Two-Hybrid System Techniques , Tyrosine/chemistry
8.
J Biol Chem ; 280(1): 225-35, 2005 Jan 07.
Article in English | MEDLINE | ID: mdl-15513915

ABSTRACT

Ras guanine-releasing factor 1 (RasGrf1), a guanine nucleotide exchange factor for members of the Ras and Rho family of GTPases, is highly expressed in the brain. It is regulated by two separate mechanisms, calcium regulation through interaction with its calcium/calmodulin-binding IQ domain and serine and tyrosine phosphorylation. RasGrf1 is activated downstream of G-protein-coupled receptors and the non-receptor tyrosine kinases, Src and Ack1. Previously, we demonstrated a novel interaction between the intracellular domain of the nerve growth factor-regulated TrkA receptor tyrosine kinase and an N-terminal fragment of RasGrf1. We now show that RasGrf1 is phosphorylated and interacts with TrkA, -B, and -C in co-transfection studies. This interaction and phosphorylation of RasGrf1 is dependent on the HIKE domain of TrkA (a region shown to interact with pleckstrin homology domains) but not on any of the phosphotyrosine residues that act as docking sites for intracellular signaling molecules such as Shc and FRS-2. The PH1 domain alone of RasGrf1 is sufficient for phosphorylation by the TrkA receptor. A potential role for Trk activation of RasGrf1 is suggested through transfection studies in PC12 cells in which RasGrf1 significantly increases neurite outgrowth at low doses of neurotrophin stimulation. Notably, this neurite outgrowth is dependent on an intact HIKE domain, as nnr5-S10 cells expressing a TrkA HIKE domain mutant do not exhibit potentiated neurite outgrowth in the presence of RasGrf1. These studies identify RasGrf1 as a novel target of neurotrophin activation and suggest an additional pathway whereby neurotrophin-stimulated neurite outgrowth may be regulated.


Subject(s)
Nerve Growth Factors/metabolism , Neurites/physiology , Receptor, trkA/metabolism , ras-GRF1/metabolism , Animals , Binding Sites , Cell Enlargement , Humans , Mice , Neurites/ultrastructure , PC12 Cells , Phosphorylation , Protein Binding , Protein Structure, Tertiary , Rats , Receptor, trkB/metabolism , Signal Transduction , Tyrosine/metabolism
9.
J Cell Biol ; 164(6): 851-62, 2004 Mar 15.
Article in English | MEDLINE | ID: mdl-15024033

ABSTRACT

We provide the first characterization of a novel signaling adapter, Nesca, in neurotrophic signal transduction. Nesca contains a RUN domain, a WW domain, a leucine zipper, a carboxyl-terminal SH3 domain, and several proline-rich regions. Nesca is highly expressed in the brain, is serine phosphorylated, and mobilizes from the cytoplasm to the nuclear membrane in response to neurotrophin, but not epidermal growth factor, stimulation in a MEK-dependent process. Overexpression studies in PC12 cells indicate that Nesca facilitates neurotrophin-dependent neurite outgrowth at nonsaturating doses of nerve growth factor (NGF). Similarly, short interfering RNA studies significantly reduce NGF-dependent neuritogenesis in PC12 cells. Mutational analyses demonstrate that the RUN domain is an important structural determinant for the nuclear translocation of Nesca and that the nuclear redistribution of Nesca is essential to its neurite outgrowth-promoting properties. Collectively, these works provide the first functional characterization of Nesca in the context of neurotrophin signaling and suggest that Nesca serves a novel, nuclear-dependent role in neurotrophin-dependent neurite outgrowth.


Subject(s)
Adaptor Proteins, Signal Transducing , Adaptor Proteins, Vesicular Transport/metabolism , Carrier Proteins/metabolism , Nerve Growth Factors/metabolism , Neurites/metabolism , Nuclear Envelope/metabolism , Active Transport, Cell Nucleus/physiology , Adaptor Proteins, Vesicular Transport/genetics , Animals , Brain/cytology , Brain/metabolism , Carrier Proteins/genetics , Enzyme Activation , Humans , MAP Kinase Signaling System/physiology , Mice , Mitogen-Activated Protein Kinases/metabolism , Nerve Growth Factor/metabolism , Nuclear Localization Signals , PC12 Cells , Protein Structure, Tertiary , Rats , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Tissue Distribution
10.
J Neurotrauma ; 19(12): 1531-41, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12542855

ABSTRACT

Increased intraspinal nerve growth factor (NGF) after spinal cord injury (SCI) is detrimental to the autonomic nervous system. Autonomic dysreflexia is a debilitating condition characterized by episodic hypertension, intense headache, and sweating. Experimentally, it is associated with aberrant primary afferent sprouting in the dorsal horn that is nerve growth factor (NGF)-dependent. Therapeutic strategies that neutralize NGF may ameliorate initial apoptotic cellular responses to the injury and aberrant afferent plasticity that occurs weeks after the injury. Subsequently, the development of autonomic disorders may be suppressed. We constructed a protein including the extracellular portion of trkA fused to the Fc portion of human IgG and expressed it using a baculovirus system. Binding of our trkA-IgG fusion protein was specific for NGF with a K(d) = 4.26 x 10(-11) M and blocked NGF-dependent neuritogenesis in PC-12 cells. We hypothesized that binding of NGF in the injured cord by our trkA-IgG fusion protein would diminish autonomic dysreflexia. Severe, high thoracic SCI was induced with clip compression and the rats were treated with intrathecal infusions (4 microg/day) of trkA-IgG or control IgG. At 14 days post-SCI, the magnitude of autonomic dysreflexia was assessed. Colon distension increased mean arterial pressure (MAP) in control rats by 46 +/- 2 from 96 +/- 5 mmHg. In contrast, MAP of rats treated with trkA-IgG increased by only 30 +/- 2 mmHg. Likewise, the MAP response to cutaneous stimulation was also reduced in rats treated with trkA-IgG (20 +/- 1 vs. 29 +/- 2). In contrast, trkA-IgG treatment had no effect on heart rate responses during colon distension or cutaneous stimulation. These results indicate that treatment with trkA-IgG to block NGF suppresses the development of autonomic dysreflexia after a clinically relevant spinal cord injury.


Subject(s)
Autonomic Dysreflexia/prevention & control , Carrier Proteins/therapeutic use , Membrane Proteins/therapeutic use , Nerve Growth Factor/antagonists & inhibitors , Receptor, trkA , Spinal Cord Compression/physiopathology , Animals , Autonomic Dysreflexia/metabolism , Autonomic Dysreflexia/physiopathology , Baculoviridae/metabolism , Blood Pressure/drug effects , Carrier Proteins/pharmacology , Cells, Cultured , Colon/innervation , Colon/physiology , Disease Models, Animal , Dose-Response Relationship, Drug , Female , Heart Rate/drug effects , Immunoglobulin G/administration & dosage , Immunoglobulin G/pharmacology , Immunoglobulin G/therapeutic use , Membrane Proteins/pharmacology , Nerve Growth Factor/immunology , PC12 Cells/drug effects , Physical Stimulation , Rats , Rats, Wistar , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/pharmacology , Recombinant Fusion Proteins/therapeutic use , Spinal Cord Compression/complications
SELECTION OF CITATIONS
SEARCH DETAIL
...